ZMAT3 Human

Zinc Finger, Matrin-Type 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Roles in p53-Dependent Pathways

ZMAT3 is a direct transcriptional target of p53 and contributes to tumor suppression through multiple mechanisms:

RNA Binding and Splicing Regulation

  • Binds AU-rich elements (AREs) in mRNA 3′-UTRs to modulate stability .

  • Regulates alternative splicing of key transcripts (e.g., MDM4, MDM2) via RNA recognition, promoting nonsense-mediated decay (NMD) of oncogenic isoforms .

  • Directly interacts with FAS and 14-3-3σ mRNAs, reducing their stability and influencing apoptosis .

Tumor Suppression Mechanisms

  • In Vivo Knockout Studies:

    • Zmat3 deficiency accelerates tumorigenesis in Trp53-wildtype lung adenocarcinoma and hepatocellular carcinoma models .

    • Triple knockout (Zmat3; Puma; p21) mice develop aggressive lymphomas, highlighting synergistic roles in apoptosis and cell cycle arrest .

  • CRISPR/Cas9 Screens:

    Model SystemPhenotypeSource
    Zmat3 −/− HSPCsLeukemia/lymphoma in Puma; p21 DKO mice
    E1A;Hras G12V MEFsDominant tumor growth advantage

Cancer Prognosis

  • High ZMAT3 expression correlates with improved survival in breast and lung cancers .

  • CRISPR/Cas9 inactivation of ZMAT3 enhances proliferation in p53-wildtype cancer cells .

Senescence and Metabolic Disease

  • Hypomethylation of ZMAT3 in adipocyte precursor cells (APCs) from type 2 diabetics drives premature senescence via p53/p21 activation .

  • ZMAT3 overexpression inhibits adipogenesis, linking RNA metabolism to metabolic dysfunction .

In Vitro Studies

  • Microarray Analysis:

    • 2,447 transcripts differentially expressed upon ZMAT3 knockdown, including FAS, AKT3, and WNT1 .

    • Pathway enrichment: p53 signaling, apoptosis, and cell cycle regulation .

In Vivo Models

  • Lung Adenocarcinoma:

    • Zmat3 loss cooperates with Kras G12D to accelerate tumor growth, independent of Trp53 status .

  • Lymphomagenesis:

    • Zmat3 −/− does not accelerate c-MYC-driven lymphoma, unlike Trp53 haploinsufficiency .

Comparative Functional Analysis

FeatureZMAT3 RoleSupporting Evidence
RNA SplicingModulates exon inclusion/NMD
ApoptosisRegulates FAS mRNA stability
Cell Cycle ArrestCooperates with p21 in tumor suppression
Metabolic RegulationInhibits adipogenesis via p53/p21

Future Directions

  • Therapeutic Targeting: ZMAT3’s RNA-binding domains offer potential for small-molecule modulation in p53-active cancers.

  • Multi-Omic Integration: Proteomic and transcriptomic analyses reveal ZMAT3’s interplay with extracellular matrix remodeling and cell migration .

Product Specs

Introduction
ZMAT3 is a protein that resides in the nucleus and possesses three zinc finger domains. Both the mRNA and protein levels of ZMAT3 are elevated by the activity of wild-type p53. When artificially overexpressed, ZMAT3 can stimulate the growth of tumor cells, suggesting its involvement in the p53-dependent pathway regulating cell growth. Alternative splicing of the ZMAT3 gene results in two distinct transcript variants, which in turn encode two protein isoforms differing by a single amino acid.
Description
Recombinant human ZMAT3, expressed in E. coli, is a single polypeptide chain that is not glycosylated. It comprises 312 amino acids (residues 1-289) and exhibits a molecular weight of 34.4 kDa. A 23 amino acid His-tag is fused to the N-terminus of the ZMAT3 protein. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The ZMAT3 solution is supplied in a buffer consisting of 20mM Tris-HCl (pH 8.0), 0.2M NaCl, 2mM DTT, and 50% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To further enhance long-term stability, the addition of a carrier protein (0.1% HSA or BSA) is advised. Repeated freezing and thawing should be avoided.
Purity
The purity of the ZMAT3 protein is greater than 85% as determined by SDS-PAGE analysis.
Synonyms
Zinc Finger Matrin-Type 3, Zinc Finger Protein WIG1, P53-Activated Gene 608 Protein, P53 Target Zinc Finger Protein, WIG1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMILLQHA VLPPPKQPSP SPPMSVATRS TGTLQLPPQK PFGQEASLPL AGEEELSKGG EQDCALEELC KPLYCKLCNV TLNSAQQAQA HYQGKNHGKK LRNYYAANSC PPPARMSNVV EPAATPVVPV PPQMGSFKPG GRVILATEND YCKLCDASFS SPAVAQAHYQ GKNHAKRLRL AEAQSNSFSE SSELGQRRAR KEGNEFKMMP NRRNMYTVQN NSAGPYFNPR SRQRIPRDLA MCVTPSGQFY CSMCNVGAGE EMEFRQHLES KQHKSKVSEQ RYRNEMENLG YV

Q&A

What is ZMAT3 and what is its primary function in human cells?

ZMAT3 is an RNA-binding protein that functions as a key splicing regulator in the p53 tumor suppression pathway. Integrative analysis of the ZMAT3 RNA-binding landscape and transcriptomic profiling reveals that ZMAT3 directly modulates exon inclusion in transcripts encoding proteins of diverse functions, including the p53 inhibitors MDM4 and MDM2, splicing regulators, and components of varied cellular processes . These exons are enriched in Nonsense-Mediated Decay (NMD) signals, and accordingly, ZMAT3 broadly affects target transcript stability .

How is ZMAT3 regulated in human cells?

ZMAT3 is primarily regulated by p53, with the ZMAT3 locus being directly bound by p53 as shown through ChIP-seq data from both human and mouse cells . Specifically, researchers have identified a major p53-bound region containing a near-perfect p53 response element (RE) in the first intron of human ZMAT3 . This direct regulation makes ZMAT3 a p53-inducible gene that is consistently activated across diverse cellular contexts and stimuli. Meta-analysis of 57 human datasets revealed that ZMAT3 and CDKN1A were significantly p53-activated in all datasets from a range of human cell types, including colorectal, breast, and bone cancer cell lines as well as primary cells .

What experimental models are commonly used to study ZMAT3 function?

Several experimental models are employed to investigate ZMAT3 biology:

  • Conditional knockout mice: Zmat3 conditional knockout alleles (Zmat3^fl) with loxP sites inserted to flank critical exons (e.g., exons 4 and 5)

  • CRISPR/Cas9 genome editing: For targeted disruption of ZMAT3 or its regulatory elements

  • RNAi and CRISPR-based genetic screens: Used for unbiased identification of ZMAT3's role in tumor suppression

  • Autochthonous mouse models: Particularly Kras^G12D-driven lung and liver cancers to study ZMAT3's tumor suppressive functions

  • Multiplexed tumor assays: Using somatic genome editing and tumor barcoding (Tuba-seq Ultra) for quantitative analysis

What is the tissue distribution pattern of ZMAT3 expression in humans?

According to the Human Protein Atlas, ZMAT3 shows expression across multiple human tissues . The protein expression pattern encompasses:

  • Central nervous system: Including amygdala, basal ganglia, cerebellum, cerebral cortex, and other brain regions

  • Digestive system: Colon, duodenum, esophagus, gallbladder, liver, pancreas, and small intestine

  • Reproductive system: Endometrium, fallopian tube, ovary, prostate, and seminal vesicle

  • Respiratory system: Bronchus, lung, and nasopharynx

  • Other systems: Adipose tissue, adrenal gland, bone marrow, heart muscle, kidney, lymph node, and skin

How significant is ZMAT3 in p53-mediated tumor suppression?

ZMAT3 represents a major component of p53-mediated tumor suppression. Quantitative tumor assays using somatic genome editing revealed that ZMAT3 accounts for approximately one-third of p53 activity in suppressing lung adenocarcinoma (LUAD) . This positions ZMAT3 as one of the most important downstream effectors of the p53 tumor suppression program. Multiple genetic screens have converged on ZMAT3 as a critical p53-inducible gene for tumor suppression in both mouse models of Kras^G12D-driven lung and liver cancers and human carcinomas .

How does ZMAT3 cooperate with other p53 effectors in tumor suppression?

ZMAT3 functions in concert with other p53 target genes, most notably CDKN1A (encoding p21). In vivo CRISPR/Cas9 screens identified CDKN1A as the most potent tumor suppressor gene cooperating with ZMAT3 . Together, ZMAT3 and CDKN1A form a core program of p53-mediated tumor suppression that:

  • Regulates cell division and proliferation

  • Controls cell migration capabilities

  • Modulates extracellular matrix (ECM) organization

  • Suppresses tumorigenesis in multiple contexts

This cooperative functionality accounts for a significant portion of p53's tumor suppressive activity, with combined loss of both factors recapitulating many aspects of p53 deficiency .

What cellular pathways are affected by ZMAT3 loss in cancer cells?

Transcriptomic and proteomic analyses have revealed several key pathways affected by ZMAT3 loss:

PathwayEffect of ZMAT3 LossPotential Therapeutic Implications
Cell ProliferationEnhanced proliferation, particularly when combined with p21 lossCell cycle inhibitors
Cell MigrationIncreased migration in 3D modelsAnti-migratory compounds
ECM OrganizationDysregulation of ECM-related genesECM-targeting therapies
Integrin SignalingUpregulation of ITGA3 and ITGA6Integrin inhibitors
RNA SplicingAltered splicing of key oncogenes and tumor suppressorsSplicing modulators

At the molecular level, ZMAT3 loss dysregulates expression of genes involved in ECM organization and cell migration at both RNA and protein levels, including upregulation of ITGA3 and ITGA6, which are associated with poor patient prognosis and aggressive cellular behavior .

How can I design experiments to investigate ZMAT3's RNA binding specificity?

To characterize ZMAT3's RNA binding properties, consider these methodological approaches:

  • CLIP-seq (Cross-linking Immunoprecipitation followed by sequencing):

    • UV cross-link RNA-protein complexes in vivo

    • Immunoprecipitate ZMAT3 with specific antibodies

    • Sequence bound RNA fragments to identify binding sites

  • RNA-seq with ZMAT3 manipulation:

    • Compare transcriptomes between ZMAT3-proficient and deficient cells

    • Focus on alternative splicing events and transcript stability

    • Use computational approaches to identify enriched sequence motifs

  • Minigene splicing assays:

    • Clone specific exons and flanking introns into reporter constructs

    • Measure splicing outcomes in the presence or absence of ZMAT3

    • Mutate predicted binding sites to validate direct regulation

  • Structure-function analyses:

    • Generate domain deletion or point mutation variants of ZMAT3

    • Test their RNA binding and splicing regulatory capacities

    • Correlate structural features with functional outcomes

What methods are most effective for studying ZMAT3's impact on transcript stability?

To investigate ZMAT3's role in regulating transcript stability:

  • Actinomycin D chase experiments:

    • Treat cells with actinomycin D to block transcription

    • Measure decay rates of target transcripts over time

    • Compare stability in ZMAT3-proficient versus deficient cells

  • Pulse-chase RNA labeling:

    • Label newly synthesized RNA with 4-thiouridine

    • Chase with unlabeled media and measure labeled RNA decay

    • Calculate half-lives of ZMAT3 target transcripts

  • NMD inhibition experiments:

    • Inhibit NMD using UPF1 depletion or small molecules

    • Assess whether ZMAT3 targets are stabilized

    • Identify which ZMAT3-regulated splice variants are NMD substrates

  • Polysome profiling:

    • Fractionate cell lysates on sucrose gradients

    • Analyze distribution of ZMAT3 target transcripts

    • Determine translation efficiency correlations with stability

How should I design genetic screens to identify synthetic lethal interactions with ZMAT3 loss?

For identifying genetic vulnerabilities associated with ZMAT3 deficiency:

  • Genome-wide CRISPR-Cas9 screens:

    • Generate isogenic cell lines differing only in ZMAT3 status

    • Perform parallel screens in ZMAT3-proficient and deficient backgrounds

    • Identify genes whose loss specifically compromises viability in ZMAT3-deficient cells

  • Focused shRNA or CRISPR libraries:

    • Target pathways functionally related to ZMAT3 (RNA processing, p53 pathway)

    • Screen in multiple cell line models with different genetic backgrounds

    • Validate hits with individual knockouts and rescue experiments

  • Drug sensitivity screens:

    • Test panels of compounds against ZMAT3-proficient and deficient cells

    • Identify agents showing selective toxicity in ZMAT3-deficient contexts

    • Determine mechanisms underlying synthetic lethality

What are the best approaches for studying ZMAT3 protein interactions and complexes?

To characterize ZMAT3's interactome and protein complexes:

  • Immunoprecipitation-mass spectrometry (IP-MS):

    • Immunoprecipitate endogenous or tagged ZMAT3

    • Identify co-precipitating proteins by mass spectrometry

    • Compare interactomes under different conditions (e.g., DNA damage)

  • Proximity labeling approaches:

    • Express ZMAT3 fused to BioID or APEX2

    • Label proteins in close proximity to ZMAT3 in living cells

    • Identify labeled proteins by streptavidin pulldown and mass spectrometry

  • Co-immunoprecipitation with specific candidates:

    • Test interactions with known splicing factors or p53 pathway components

    • Validate mass spectrometry hits with targeted co-IP experiments

    • Map interaction domains through truncation mutants

  • Size exclusion chromatography:

    • Fractionate cellular lysates by size

    • Determine which fractions contain ZMAT3

    • Identify co-eluting proteins to define native complexes

How can ZMAT3 research inform therapeutic strategies for p53-deficient cancers?

ZMAT3 research offers several potential therapeutic angles:

  • Targeting downstream vulnerabilities:

    • Genes and pathways dysregulated with ZMAT3 loss, such as integrin signaling (ITGA3, ITGA6), represent potential therapeutic targets

    • ECM and cell migration pathways affected by ZMAT3 loss may be targetable

  • Exploiting synthetic lethality:

    • Identifying genes and pathways that become essential in ZMAT3-deficient contexts

    • Developing compounds that selectively target cells with compromised ZMAT3 function

  • Splicing modulation:

    • Correcting aberrant splicing events resulting from ZMAT3 loss

    • Developing splicing modulators that counteract specific ZMAT3-dependent mis-splicing events

  • Biomarker development:

    • Using ZMAT3 status or its downstream effectors as predictive biomarkers for therapy response

    • Stratifying patients based on ZMAT3 function to guide precision medicine approaches

What are the methodological considerations for analyzing ZMAT3 in clinical samples?

When studying ZMAT3 in patient specimens:

  • Sample preservation considerations:

    • RNA quality significantly impacts splicing analysis

    • FFPE samples may require specialized protocols for reliable splicing assessment

    • Fresh frozen tissues are preferable for detailed molecular analyses

  • Comprehensive analysis approaches:

    • Assess both ZMAT3 expression levels and splicing function

    • Include analysis of p53 status, as ZMAT3 function is p53-dependent

    • Examine both mRNA and protein expression patterns

  • Isoform-specific detection:

    • Develop PCR assays targeting specific ZMAT3-regulated splice junctions

    • Use RNA-seq with sufficient depth to detect alternative splicing events

    • Consider long-read sequencing for complex splicing patterns

  • Integration with clinical data:

    • Correlate ZMAT3 status with patient outcomes and therapy responses

    • Account for confounding variables, particularly p53 mutations

    • Consider ZMAT3 in the context of broader tumor molecular profiles

How might ZMAT3 function as a biomarker in cancer diagnostics or prognostics?

ZMAT3's potential as a cancer biomarker stems from:

  • Prognostic value:

    • Low ZMAT3 expression or function may indicate compromised p53 tumor suppression

    • ZMAT3-regulated splicing events could serve as surrogate markers for pathway activity

    • Combined assessment of ZMAT3 and CDKN1A may provide robust prognostic information

  • Predictive utility:

    • ZMAT3 status might predict response to therapies targeting RNA splicing

    • Specific ZMAT3-regulated splice variants could indicate sensitivity to particular treatments

    • ZMAT3 deficiency might correlate with response to drugs targeting synthetic lethal interactions

  • Implementation approaches:

    • Develop RNA-based diagnostics targeting ZMAT3-regulated splice junctions

    • Create antibodies specific to ZMAT3 protein or its key targets

    • Design multiplexed assays examining ZMAT3 alongside other p53 pathway components

How can I overcome technical challenges in studying ZMAT3's splicing targets?

Common challenges and solutions include:

ChallengeTechnical Solution
Distinguishing direct from indirect targetsCombine CLIP-seq binding data with splicing outcome analysis
Low abundance of alternatively spliced isoformsUse targeted amplification or junction-specific qPCR
Functional validation of splicing eventsCRISPR-based editing of specific splice sites
Cell type-specific regulationStudy multiple cellular contexts to identify core targets
Temporal dynamics of regulationConduct time-course experiments after p53 activation
Distinguishing splicing from stability effectsAnalyze pre-mRNA and mature mRNA levels separately
Protein-level confirmation of splicing changesDevelop isoform-specific antibodies

How should I interpret contradictory data regarding ZMAT3 function across different cancer types?

When facing inconsistent findings:

  • Consider context-dependent factors:

    • p53 status impacts ZMAT3 function and significance

    • Tissue-specific expression of ZMAT3 cofactors may alter outcomes

    • Cancer stage and molecular subtype may influence ZMAT3 dependency

  • Evaluate methodological differences:

    • Acute versus chronic ZMAT3 loss may yield different phenotypes

    • In vitro versus in vivo models often show different dependencies

    • RNAi versus genetic knockout approaches may have different specificities

  • Validation strategies:

    • Use multiple independent approaches to confirm findings

    • Test hypotheses across diverse model systems

    • Consider genetic background differences that might influence outcomes

    • Correlate experimental findings with patient data when possible

What controls are essential when studying ZMAT3's role in RNA processing?

Critical controls include:

  • Genetic controls:

    • ZMAT3 rescue experiments to confirm specificity

    • p53-null controls to distinguish p53-dependent and independent effects

    • CDKN1A knockout comparisons to differentiate from other p53 effectors

  • Experimental controls:

    • RNA binding mutants that maintain protein expression but lack function

    • Off-target assessment for RNAi or CRISPR approaches

    • Time-course experiments to capture dynamic regulatory events

  • Analytical controls:

    • Global splicing pattern analysis to identify specific versus general effects

    • Assessment of transcription rate versus RNA stability

    • Comparison with other RNA binding protein perturbations

Product Science Overview

Introduction

Zinc Finger, Matrin-Type 3 (ZMAT3) is a protein encoded by the ZMAT3 gene in humans. This protein is characterized by the presence of three zinc finger domains and a nuclear localization signal . Zinc finger proteins are a large family of transcription factors that play crucial roles in gene regulation by binding to DNA sequences.

Gene and Protein Structure

The ZMAT3 gene is located on chromosome 3 and encodes a protein that contains three zinc finger domains . These domains are critical for the protein’s ability to bind to DNA and regulate gene expression. The protein also contains a nuclear localization signal, which directs it to the nucleus where it can interact with DNA .

Function and Mechanism

ZMAT3 is upregulated by the tumor suppressor protein p53, which is known for its role in preventing cancer development . The overexpression of ZMAT3 has been shown to inhibit tumor cell growth, suggesting that it may play a role in the p53-dependent growth regulatory pathway . This indicates that ZMAT3 could be involved in the regulation of cell growth and apoptosis, contributing to the suppression of tumor formation.

Biological Significance

The ZMAT3 protein is a bona fide target gene of p53/TP53 and may contribute to TP53-mediated apoptosis by regulating TP53 expression and its translocation to the nucleus and nucleolus . This highlights the importance of ZMAT3 in the p53 signaling pathway, which is a critical pathway for maintaining cellular homeostasis and preventing tumorigenesis.

Clinical Implications

Given its role in the p53-dependent growth regulatory pathway, ZMAT3 has potential clinical implications in cancer research. Its ability to inhibit tumor cell growth makes it a potential target for cancer therapies. Further research is needed to fully understand the mechanisms by which ZMAT3 regulates cell growth and apoptosis and to explore its potential as a therapeutic target.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.